WO2016162675A1 - Therapeutic compositions and methods of use for treating cancer - Google Patents

Therapeutic compositions and methods of use for treating cancer Download PDF

Info

Publication number
WO2016162675A1
WO2016162675A1 PCT/GB2016/050973 GB2016050973W WO2016162675A1 WO 2016162675 A1 WO2016162675 A1 WO 2016162675A1 GB 2016050973 W GB2016050973 W GB 2016050973W WO 2016162675 A1 WO2016162675 A1 WO 2016162675A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
tumor
virus
cancer
cell
Prior art date
Application number
PCT/GB2016/050973
Other languages
English (en)
French (fr)
Inventor
Uri Galili
Michael WESTBY
Original Assignee
Agalimmune Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to BR112017021700-7A priority Critical patent/BR112017021700A2/pt
Priority to KR1020177027601A priority patent/KR102544032B1/ko
Priority to RU2017134519A priority patent/RU2720984C2/ru
Priority to EP16716896.2A priority patent/EP3280798B1/en
Priority to CA2981925A priority patent/CA2981925A1/en
Priority to JP2017552992A priority patent/JP2018512150A/ja
Priority to MX2017012867A priority patent/MX2017012867A/es
Priority to ES16716896T priority patent/ES2743952T3/es
Application filed by Agalimmune Limited filed Critical Agalimmune Limited
Priority to US15/564,774 priority patent/US20180104288A1/en
Priority to CN201680020049.0A priority patent/CN107787364A/zh
Publication of WO2016162675A1 publication Critical patent/WO2016162675A1/en
Priority to IL254842A priority patent/IL254842B/en
Priority to HK18110254.0A priority patent/HK1251009B/zh
Priority to US16/698,682 priority patent/US20200155626A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/761Adenovirus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/768Oncolytic viruses not provided for in groups A61K35/761 - A61K35/766
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/45Transferases (2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0031Rectum, anus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0034Urogenital system, e.g. vagina, uterus, cervix, penis, scrotum, urethra, bladder; Personal lubricants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0043Nose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1048Glycosyltransferases (2.4)
    • C12N9/1051Hexosyltransferases (2.4.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y204/00Glycosyltransferases (2.4)
    • C12Y204/01Hexosyltransferases (2.4.1)
    • C12Y204/01087N-Acetyllactosaminide 3-alpha-galactosyltransferase (2.4.1.87), i.e. alpha-1,3-galactosyltransferase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10332Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10371Demonstrated in vivo effect
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16611Simplexvirus, e.g. human herpesvirus 1, 2
    • C12N2710/16632Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16611Simplexvirus, e.g. human herpesvirus 1, 2
    • C12N2710/16641Use of virus, viral particle or viral elements as a vector
    • C12N2710/16643Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16611Simplexvirus, e.g. human herpesvirus 1, 2
    • C12N2710/16671Demonstrated in vivo effect
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y204/00Glycosyltransferases (2.4)
    • C12Y204/01Hexosyltransferases (2.4.1)
    • C12Y204/01057Phosphatidylinositol alpha-mannosyltransferase (2.4.1.57)

Definitions

  • the present invention relates to compositions and methods for treating cancer. More specifically, the present invention relates to compositions of engineered oncolytic viruses for administration to a subject with cancer that specifically lyse tumor cells and actively target tumor cells and cell debris to antigen presenting cells, in order to generate antitumor immunity.
  • Tumors may develop in cancer patients because the immune system fails to detect tumor cells as cells that ought to be destroyed.
  • Tumor cells express autologous tumor antigens in a large proportion of cancer patients. These autologous tumor antigens, also called “neoantigens", may elicit a protective anti-tumor immune response.
  • Tumor cells, or tumor cell membranes have to be internalized by antigen presenting cells in order to induce the development of an anti-tumor immune response.
  • the immune system in many cancer patients displays an "ignorance" toward the tumor antigens that is associated with early development of the tumor in a "stealthy” way, so that the tumor is effectively "invisible” to antigen presenting cells (Pardoll, 2000; Clin Immunol. 95:S44- 49, and Dunn et al, 2002; Nat Immunol; 3: 991-8).
  • tumor microenvironment and local cytokine milieu are often suppressive toward immune function and can actively induce immune cell anergy and death
  • Such an immune response will destroy any untreated lesions (for example, those that cannot be accessed for treatment nor removed by surgery) and results in immune mediated detection, regression, and/or destruction of micrometastases which cannot be detected visually and are not detectable by imaging.
  • the size of a tumor impedes the efficacy of a circulating anti-tumor immune response in a timely manner.
  • Surgical resection or other means such as intratumoral injection of compositions are often necessary to reduce the size of a tumor in addition to harnessing an active antitumor immunity.
  • oncolytic viruses have been developed that are useful in selectively killing tumor cells by lytic replication and thereby reducing tumor size (Liu et a/., World J Gastroenterol.
  • HSV herpes simplex virus
  • GM-CSF granulocyte macrophage colony stimulating factor
  • Induction of a protective anti-tumor immune response requires uptake of the tumor cells and cell components by antigen-presenting cells, then processing of tumor antigens and their transportation by antigen presenting cells to the draining lymph nodes.
  • the immunogenic tumor antigen peptides are presented by antigen presenting cells in association with class I or class II MHC molecules for the activation of tumor specific CD8 + and CD4 + T cells, respectively. Only after these T cells are activated by the processed and presented tumor antigen peptides, these lymphocytes proliferate, leave the lymph nodes and circulate in the body to seek and destroy metastatic tumor cells expressing the relevant tumor antigens. Therefore, eliciting an effective anti-tumor immune response requires effective and active targeting of tumor cells to antigen presenting cells.
  • an oncolytic virus comprising a nucleic acid encoding a hexosyl transferase enzyme.
  • a pharmaceutical composition comprising the oncolytic virus as defined herein in combination with a pharmaceutically acceptable carrier.
  • a method of treating an individual with a neoplasm which comprises the steps of:
  • a method of treating cancer comprising administering a therapeutically effective amount of the oncolytic virus as defined herein to a patient suffering from cancer or having a neoplasm or tumor in need of treatment.
  • FIGURE 1 Provides a schematic showing a series of cellular events relevant to the present methods. These events include A) administration of engineered viral compositions, B) infection, replication, expression of alpha 1 ,3-galactosyltransferase, and alpha-gal epitopes, and C) oncolysis and recruitment of antigen presenting cells at the tumor site.
  • FIGURE 2 Provides a schematic showing a tumor cell infected with oncolytic virus containing alpha 1 ,3-galactosyl transferase and expressing alpha-gal epitopes on its cell surface prior to lysis.
  • Alpha-gal-labelled membrane fragments are internalized by antigen-presenting cells.
  • Tumor cells contain tumor associated antigens (TAA) in the cytoplasm and on the cell membrane, which are unique to the tumor and to each individual patient. These TAA are presented on the cell membrane as ( ⁇ , ⁇ , ⁇ ) and as protein molecules inside the cell (spirals and wavy lines).
  • TAA tumor associated antigens
  • the oncolytic virus (filled circles with 1 1 external spokes) introduced into the tumor lesion contains the alpha 1 ,3- galactosyltransferase gene inserted into its genome.
  • the oncolytic virus infecting the tumor cells introduces the alpha 1 ,3-galactosyltransferase gene into the tumor cells. This gene is transcribed and translated within the infected cell resulting in the production of the alpha 1 ,3-galactosy transferase (a1 ,3GT) enzyme. This enzyme synthesizes alpha- gal epitopes on cell surface glycoproteins, glycolipids and proteoglycans.
  • binding of the natural anti-Gal antibody to these alpha-gal epitopes activates the complement system and thus generates chemotactic factors in the form of complement cleavage peptides that recruit antigen presenting cells into the treated tumor lesion.
  • the tumor cells are lysed by the oncolytic virus and by anti-Gal binding to the alpha-gal epitopes on the cell membranes by complement-dependent cytotoxicity and by antibody-dependent cellular cytotoxicity (ADCC).
  • the dead cells and fragmented cell membranes coated with the anti-Gal antibody (that is bound to the alpha-gal epitopes), are internalized by antigen presenting cells (APC) via interaction between the Fc portion of the anti-Gal antibody coating the cells, or cell membranes, and Fey receptors on the APC.
  • APC antigen presenting cells
  • TAAs internalized as a result of this uptake are processed by the APC, and the TAA peptides are then presented on the APC cell membrane in association with MHC molecules.
  • the APCs migrate to the draining lymph nodes where the presented TAA peptides bind to the T cell receptors of tumor specific T cells. These T cells are activated as a result of this interaction.
  • the activated tumor specific T cells proliferate and leave the lymph nodes in order to circulate, detect and destroy metastatic tumor cells.
  • FIGURE 3 Provides a schematic timeline for appearance of alpha-1 ,3 galactosyltransferase (a1 ,3GT) and alpha-gal epitopes in HeLa cells transduced by adenovirus containing the a1 ,3GT gene (AdaGT).
  • FIGURE 4 Provides a graph showing expression of alpha-gal epitopes on mouse B16 melanoma cells transduced with AdaGT, as indicated by binding of
  • B16 A daGT cells i.e. B16 cells transduced by AdaGT [1x10 10 infectious units (IU)/ml]
  • the lectin is coupled to fluorescein (FITC).
  • Thin line histogram with dotted area- B16 A dcont cells i.e. melanoma cells transduced with adenovirus containing no inserted genes
  • thick line histogram- B16 A daGT cells i.e. melanoma cells transduced with adenovirus containing no inserted genes.
  • FIGURE 5 Provides a graph showing protection of alpha 1 ,3- galactosyltransferase knockout mice from B16 tumor challenge using B16 Ada GT vaccine.
  • Alpha 1 ,3-galactosyltransferase knockout mice producing anti-Gal were vaccinated with 2x10 6 irradiated B16 cells expressing alpha-gal epitopes ( ⁇ 16 ⁇ ⁇ )( ⁇ ), or with irradiated B16 cells transduced with control adenovirus (B16 A dcont) (O). This immunization was repeated after one week. The mice were challenged, one week later, with 0.2 x 10 6 live B16 cells (A), or 0.5 x 10 6 live B16 cells (B).
  • mice were monitored every day for 2 months for tumor growth. Results are presented as percentage of mice remaining tumor-free at various days post tumor challenge. Results are of 22 mice in the experimental group and 21 in the control group in (A) and in separate experiment of 21 mice in the experimental group and 18 in the control group in (B).
  • FIGURE 6 Provides the genomic structures of CRAd-aGT (A) and CRAd-GFP (B).
  • the Ad5 genomic sequence was modified to express the fiber knob of Ad3.
  • the E3 or E1 sequences were modified by deletion or replacement with the nucleotide sequences of the a1 ,3GT or GFP genes, respectively.
  • FIGURE 7 Quantification of human cancer cell viability after infection with CRAd- aGT.
  • A549 and A375 cells were seeded in 96-well plates, infected with a serial titration of virus starting from 1.2x10 11 virus particles (VP)/ml CRAd-aGT or 9.7x10 10 VP/ml CRAd-GFP control virus and incubated for 72 hours.
  • Cell viability was quantified using a luminescent cell viability assay Cell Titre Glo (Promega).
  • FIGURE 8 Visualization of human cancer cell viability after infection with CRAd- aGT.
  • A549 (lung carcinoma) and A375 (melanoma) cells were seeded in 24-well plates, infected with 3.6x10 7 virus particles (VP)/ml CRAd-aGT or 1.9x10 10 VP/ml CRAd-GFP control virus and incubated for 72 hours. Visualization was performed using light microscopy at 10x magnification.
  • FIGURE 9 Analysis of anti-Gal binding to human cancer cells after infection with CRAd-aGT.
  • A549 and A375 cells were infected with either 1.44x10 6 virus particles (VP)/ml CRAd-aGT or 1.16x10 6 VP/ml CRAd-GFP control virus. The cells were analyzed for anti-Gal binding and GFP expression by flow cytometry.
  • VP virus particles
  • FIGURE 9 Analysis of anti-Gal binding to human cancer cells after infection with CRAd-aGT.
  • an oncolytic virus comprising a nucleic acid encoding a hexosyl transferase enzyme.
  • the present invention relates to compositions and methods to combine oncolytic virus- induced lysis of tumor cells with active immune-mediated targeting and recruitment of antigen presenting cells (APCs) to a tumor site.
  • Engineered oncolytic viruses that express a hexosyltransferase, such as alpha 1 ,3-galactosyltransferase, will modify the glycosylation of infected cells such that they express alpha-gal epitopes on cell membranes prior to virus-induced lysis.
  • the tumor cells are subsequently lysed by the virus, the cell membrane fragments will be opsonized by natural anti-Gal antibodies that bind the alpha-gal epitopes to form immune complexes.
  • the anti-Gal antibodies will enhance the phagocytosis of the tumor cell fragments by APCs through the interaction between the Fc region of the anti-Gal antibodies and Fey receptors (FcyR) on the APCs.
  • FcyR Fey receptors
  • intact tumor cells that express alpha-gal epitopes after virus infection will also be opsonized by anti-Gal and taken up by APCs via FcyRs. Uptake of antigen via FcyRs results in activation and maturation of APCs, which process the tumor antigens for presentation on MHC molecules and migrate to the draining lymph nodes, where they present the tumor antigens to T cells. By this process a protective immune response is generated against the patient's own tumor antigens.
  • complement results in: release of chemotactic peptides that recruit APCs to the tumor; opsonization of intact tumor cells and tumor cell membrane fragments with complement C3b molecules; complement-mediated lysis of intact tumor cells. Intact tumor cells and tumor cell fragments opsonized with complement C3b molecules are bound and internalized by APCs via the interaction between C3b and complement receptors on the APC.
  • the present invention thus provides an improved treatment of various types of cancer, and even the treatment of such types that have as of yet been thought to be not curable is possible.
  • the present invention is directed to compositions and methods of using oncolytic viruses that have been modified to deliver a heterologous nucleic acid sequence encoding a glycosyl transferase gene, such as alpha 1 ,3-galactosyltransferase.
  • a heterologous nucleic acid sequence encoding a glycosyl transferase gene, such as alpha 1 ,3-galactosyltransferase.
  • the premise being that the virus specifically infects tumor cells, which then produce and express the functional alpha 1 ,3-galactosyltransferase enzyme.
  • the alpha 1 ,3-galactosyltransferase enzyme subsequently produces and integrates alpha-gal epitopes on the surface of the tumor cells.
  • alpha-gal epitopes on the tumor cell membrane occurs prior to cell lysis induced by the oncolytic virus, such that the subsequent cellular membrane debris is labeled with alpha-gal epitopes.
  • binding of alpha-gal labeled tumor cell fragments by the natural anti-Gal antibody activates the complement system (as do most antigen/antibody interactions) and generates complement cleavage chemotactic factors that recruit APCs to the treated tumor.
  • the tumor cells and tumor cell membrane fragments that express alpha-gal epitopes are opsonized by anti-Gal and actively targeted for phagocytosis by the APCs recruited to the tumor.
  • tumor cell membranes are more efficiently phagocytosed by APCs because the tumor cell fragments or intact tumor cells express alpha-gal epitopes, which subsequently bind anti-Gal antibodies.
  • the interaction between the Fc portion of the anti-Gal antibodies coating the tumor cell fragments/intact tumor cells and the FcyRs on APCs stimulate the APCs to internalize, process and present the antigens to T cells.
  • the activation of complement and the subsequent opsonization of the tumor cell fragments and intact tumor cells with complement protein C3b enables APCs bearing complement receptors to effectively internalize, process and present tumor antigens to T cells.
  • the present methods are counter intuitive because one would not presumably seek to modify tumor cell surface glycosylation with a replication competent oncolytic virus to target the cell membrane fragments and intact cells for uptake by APCs.
  • the expression and cell membrane integration of alpha-gal epitopes prior to virus- induced cell lysis provides a method to label tumor cell membrane fragments for APC recruitment and active uptake by APCs, inducing a protective anti-tumor immune response as compared to the less efficient methods previous described. While not to be limited by any specific mechanism, schematics are provided in Figures 1 and 2 which show one possible mechanism by which it is believed that the claimed compositions and methods have therapeutic action.
  • Embodiments of the present disclosure will employ, unless otherwise indicated, techniques of medicine, organic chemistry, biochemistry, molecular biology,
  • the enzyme is a galactosyl transferase enzyme. In a further embodiment, the enzyme is an alpha 1 ,3-galactosyltransferase enzyme.
  • compositions useful herein comprise an oncolytic virus and a nucleic acid sequence encoding a galactosyl transferase and optionally a pharmaceutically acceptable carrier useful for administration.
  • a pharmaceutical composition comprising the oncolytic virus as defined herein in combination with a pharmaceutically acceptable carrier.
  • the virus is engineered to be oncolytic. In an alternative
  • the virus is naturally oncolytic.
  • the oncolytic virus comprises a recombinant binding domain specific for a tumor stem cell marker.
  • the virus is replication restricted and only lyses cancer cells leaving non-cancer cells.
  • Replication-restrictive oncolytic viruses are particularly useful for the compositions and methods described herein. Such viruses only replicate in and kill cancer cells. They may additionally encode a heterologous gene (or genes) that encodes for a protein, which has additional anti-tumor properties.
  • the term oncolytic virus is meant to comprise any virus that infects/enters and lyses cancer cells. The ideal oncolytic virus efficiently kills a clinically relevant fraction of the patient's cancer cells by direct cytolysis with a minimal destruction of non-neoplastic tissue.
  • Oncolytic viruses are preferred (from a safety perspective) if they naturally are not pathogenic (e.g. naturally do not infect humans) or only cause mild disease in humans (e.g. adenoviruses cause flu-like symptoms). Viruses that have been used successfully in approved vaccines and have been administered to thousands or millions of people (e.g. small pox vaccine) are also preferred for this reason. If a virus is pathogenic in humans and is linked to significant disease (e.g. neurotoxicity associated with some herpes virus strains) then it is preferred to make multiple deletions or mutations in the viral genome to render them specific for cancer cells and reduce the risk that a single genetic recombination event with an endogenous virus leads to a fully pathogenic strain.
  • significant disease e.g. neurotoxicity associated with some herpes virus strains
  • Oncolytic viruses derived from many different types of viruses have been described by Liu et al. (Liu et al., Nature Clinical Practice Oncology 4: (2) 101-1 17, 2007).
  • HSV herpes simplex virus
  • VV vaccinia virus
  • paramyxoviruses such as measles virus (MeV), Newcastle disease virus (NDV) or rhabdoviruses like vesicular stomatitis virus (VSV), are most prominent.
  • MeV measles virus
  • NDV Newcastle disease virus
  • VSV vesicular stomatitis virus
  • genetic engineering can further improve safety and efficacy of oncolytic viruses.
  • a key feature of replication restrictive oncolytic virus vectors that encode a heterologous gene is that the initial dose of heterologous gene can be amplified in vivo many fold by replication in, and release from, cancer cells within the injected lesion(s). This is taught, for example, by Liu et al (Liu et al., 2003), who describes the features of a mutant herpes simplex virus-1 (HSV-1 ) vector that encodes granulocyte macrophage-colony stimulating factor (GM-CSF). Collectively, these cancer therapies have been termed "armed therapeutic vaccines" (Bauzon & Hermiston, 2014).
  • Viruses may be derived from well-characterized lab-adapted strains that have been maintained by multiple serial passages through immortalized cell lines in tissue culture.
  • human tumors in patients grow more slowly than do immortalized cell lines in optimized culture conditions, and may have other environmental and development factors that limit the rate of metabolism and cell division (such as the hypoxic environment that can exist at the center of a large tumor mass, a result of an insufficient nutrient supply). Therefore, it is preferable to select a virus for optimal growth in vivo (in situ) rather than based on its growth characteristics in tissue culture.
  • an HSV-1 strain derived from clinical isolate JS1 obtained from a patient and having only undergone a short time of propagation in tissue culture
  • JS1 obtained from a patient and having only undergone a short time of propagation in tissue culture
  • oncolytic viruses for treatment of cancer patients (Liu, 2003).
  • An important consideration in selecting a clinical isolate is that optimal conditions can be identified such that it can be propagated in vitro, which is necessary for scale up and manufacture of the clinical material. Mutations to the genome are envisaged that improve the replication of virus in tissue culture without impairing its growth in tumors in vivo.
  • a virus vector In addition to selecting a virus vector based on its ability to replicate in and lyse tumor tissue in patients, additional mutations to the genome are envisaged that further improve its ability to replicate in vivo.
  • two genes are deleted from clinical HSV-1 strain, JS1 (Liu, 2003).
  • the first deletion, ICP34.5 restricts virus replication to cancer cells;
  • the second, ICP47 results in the early expression of US1 1 , which encodes a viral protein that blocks phosphorylation of host protein, PKR.
  • PKR is a key component of the innate immune response to viral infection - upon phosphorylation it shuts down protein translation in infected cells and limits viral replication. Therefore, the in vivo replication of the mutant JS1 ICP34.5-/ICP47- strain is enhanced relative to the wild type (non- mutated) strain.
  • the oncolytic virus is an oncolytic replication competent adenovirus, vaccinia virus, herpes virus, reovirus, measles virus or Newcastle disease virus.
  • the virus is an RNA or DNA based virus of human or non- human origin, such as adenovirus, herpesvirus, vaccinia virus, measles virus, Newcastle Disease Virus, autonomous parvoviruses, vesicular stomatitis virus (VSV) or reovirus.
  • adenovirus such as adenovirus, herpesvirus, vaccinia virus, measles virus, Newcastle Disease Virus, autonomous parvoviruses, vesicular stomatitis virus (VSV) or reovirus.
  • VSV vesicular stomatitis virus
  • Table 1 Exemplary oncolytic viruses shown in Table 1 are currently in clinical trials and may be modified as described herein for use in the therapeutic methods provided.
  • the oncolytic virus is an enveloped virus derived from the virus families herpesviridae, poxviridae, rhabdoviridae, or paramyxoviridae, preferably from the Paramyxovi
  • Herpesvirus G207 Glioma Herpesvirus G207 Glioma
  • oncolytic viruses are genetically modified to confer tumor-specific replication (adenovirus, herpesvirus, vaccinia virus) while others are naturally tumor-specific viruses (reovirus, Newcastle disease virus, autonomous parvovirus, certain measles strains, and vesicular stomatitis virus (VSV)).
  • adenovirus adenovirus, herpesvirus, vaccinia virus
  • reovirus Newcastle disease virus, autonomous parvovirus, certain measles strains, and vesicular stomatitis virus (VSV)
  • VSV vesicular stomatitis virus
  • the oncolytic virus is an adenovirus.
  • Human adenoviruses are non- enveloped, double-stranded DNA viruses of about 30-38 kB.
  • a key feature of adenoviruses is that they encode a number of viral proteins that inhibit critical host regulatory proteins. Some of these host regulatory proteins (such as p53) are defective in many cancers. Therefore, deletion of the viral gene renders the mutant viruses selective for replication in cancers with the defective regulatory host protein.
  • mutation of the adenovirus E1 B-55kD gene a p53-inhibitory protein, (for example in the virus dl1520), is selective for tumors that have limited or no p53 function.
  • p53 function is lost in most human cancers through various mechanisms including gene mutation, overexpression of p53-binding inhibitors (e.g. mdm2, human papillomavirus
  • deletions in the E1 a conserved region 2 have defective retinoblastoma (RB) protein binding, and these mutants are being evaluated for use against tumors with RB pathway abnormalities (for example in the virus d/922/947).
  • RB retinoblastoma
  • Another approach to obtain cancer-cell selective replication of adenoviruses is to control expression of the viral E1a gene product using a promoter that is specific for the tissue or cancer of interest. For example, this has been applied to oncolytic adenoviruses targeting prostate cancer (by genetic modification of the genome to introduce the prostate specific antigen (PSA) promoter/enhancer upstream of the E1 a gene.
  • PSA prostate specific antigen
  • the virus is a conditionally replicating adenovirus (CRAd), in which replication is restricted to tumor cells.
  • CRAds have been demonstrated to selectively replicate in cancer cells, causing their lysis and death (Fueyo et al., 2000; Kanno et al., 2012; Bramante et al., 2015).
  • CRAds have been generated using adenovirus 5 (Ad5) that has been genetically modified to express the fiber knob of adenovirus 3 (Ad3), to create an Ad5/3 chimeric virus (Kanerva et al. , 2003; Kim et al., 2013).
  • the CRAd is an Ad5/3 chimeric virus.
  • the chimeric Ad5/3 virus is able to bind to cells and gain entry using CD46, the Ad3 receptor, rather than the coxsackie-adenovirus receptor (CAR), which bound by Ad5.
  • CD46 coxsackie-adenovirus receptor
  • This is advantageous as many tumors have variable expression of CAR, whereas all nucleated cells express CD46 (Ulasov et al., 2006).
  • the virus is rendered conditionally replicative by introducing a 24-base pair deletion ( ⁇ 24) in constant region 2 (CR2) of the viral immediately early (E1a) gene (Kanerva et a/., 2003).
  • the Ad5/3 chimeric virus additionally comprises a 24-base pair deletion ( ⁇ 24) in constant region 2 (CR2) of the viral immediately early (E1 a) gene, herein known as the Ad5/3-A24 CRAd.
  • This mutation results in a viral E1 a protein that is unable to bind retinoblastoma (Rb) protein for induction of S phase within the cell (Fueyo et a/., 2000). Therefore, the virus is unable to replicate in non-dividing normal cells, but efficiently replicates in cells that have an inactive Rb/p16 pathway, a phenotype indicated to be shared by all cancers (Sherr, 1996).
  • Chimeric, conditionally replicating Ad5/3-A24 CRAd can be further modified by the substitution of the adenovirus early region (E3) gene with a transgene, which results in an Ad5/3-A24 CRAd that specifically produces the transgene during replication (Koski et a/., 2010; Kanerva et a/., 2013; Bramante et a/., 2015).
  • E3 adenovirus early region
  • the chimeric, conditionally replicating Ad5/3-A24 CRAd has been further modified by the substitution of the adenovirus early region (E3) gene with an alpha 1 ,3-galactosy transferase transgene, which results in an Ad5/3-A24 CRAd that specifically produces alpha 1 ,3- galactosyltransferase during replication.
  • the Ad5/3-A24 CRAd additionally comprises an alpha 1 ,3-galactosyltransferase transgene, herein known as CRAd-aGT.
  • the oncolytic virus is a herpes virus.
  • Methodology is presented herein in Examples 4-7 which demonstrates how positive results may be achieved using alpha-1 ,3 galactosyltransferase containing herpes virus.
  • Herpes virus (HSV) is an enveloped, double-stranded DNA virus of about 152 kbp, and has a natural tropism for neuronal and mucosal cells. HSV has been suggested to be of use both as a gene delivery vector in the nervous system and elsewhere and for the oncolytic treatment of cancer. In both applications the virus must however be disabled such that it is no longer pathogenic but such that it can still enter cells and perform the desired function.
  • Viruses deleted for only ICP34.5 have been shown to replicate in many tumor cell types in vitro and to selectively replicate in artificially induced brain tumors in mice while sparing surrounding tissue. Early stage clinical trials have also shown their safety in man. Examples of such viruses and uses are described in PCT publications WO 2001/053506, WO 98/004726, WO 98/051809 and WO 99/060145.
  • the oncolytic virus is an HSV1 virus.
  • One exemplary oncolytic virus strain is HSV1 strain JS 1 deposited at the European Collection of Cell Cultures
  • the oncolytic virus is a vaccinia virus.
  • Vaccinia virus is an enveloped, double-stranded DNA virus of about 200 kB. This virus replicates in the cytoplasm, bringing with its infectious particle the enzymes required for DNA replication and transcription of its genes.
  • Vaccinia virus has many attributes that make it a good oncolytic virus. It has a large genome with the capacity to accommodate multiple foreign genes, it has a broad host range, it replicates rapidly, and it can easily be recombined for making viral mutants. A good safety record has been established through use of vaccinia virus as a smallpox vaccine. iv) Measles virus
  • the virus is a measles virus (MeV) or a vaccine strain of MeV such as the Edmonston strain (MeV E dm).
  • MeV utilizes two envelope glycoproteins (the fusion protein (F) and the hemagglutinin protein (H)) to gain entry into the target cell.
  • F fusion protein
  • H hemagglutinin protein
  • Protein F is a type I transmembrane protein
  • protein H is a type II transmembrane domain, i.e. its amino-terminus is exposed directly to the cytoplasmic region. Both proteins thus comprise a transmembrane and a cytoplasmic region.
  • One known function of the F protein is mediating the fusion of viral membranes with the cellular membranes of the host cell.
  • H protein Functions attributed to the H protein include recognizing the receptor on the target membrane and supporting F protein in its membrane fusion function.
  • the direct and highly efficient membrane fusion at the cellular surface membrane is a particular property of measles virus and the morbilliviruses, thus distinguishing themselves from many other enveloped viruses that become endocytosed and will only fuse upon pH drop upon endocytosis.
  • Both proteins are organized on the viral surface in a regular array of tightly packed spikes, H tetramers, and F trimers (Russell et al., Virology 199:160-168, 1994).
  • the Edmonston strain of MeV uses a single protein as its main receptor, namely, the protein known to be the regulator of complement activation factor, CD46 (Gerlier et al., Trends Microbiol. 3:338-345, 1995). CD46 is expressed on all nucleated human cells. Most clinical isolates of measles virus, however, cannot effectively use CD46 as a receptor.
  • Human SLAM signal lymphocyte-activation molecule; also known as CDw150
  • CDw150 is a recently discovered membrane glycoprotein that is expressed on some T and B cells, and was also found to act as a cellular receptor for MeV, including the Edmonston strain (Tatsuo et al., Nature 406(6798):893-7, 2000). The precise biological functions and interactions of the MeV H and F proteins remain largely unclear.
  • oncolytic viruses in clinical development for cancer that can be modified to include a hexosyl transferase encoding nucleic acid sequence such as alpha1 ,3-galactosyltransferase.
  • oncolytic viruses include Ad-mda7 (p53 Inc), Ad-p53 (p53 Inc.), CG-0070 (Cold Genesys, Inc.), DNX-2401 (DNAtrix, Inc.), DWP-418 (Daewoong Pharmaceutical), HSV expressing hulL-12 (Univ. Alabama Birmingham), G-47 Delta (Univ.
  • viruses of the invention infect and replicate in tumor cells, subsequently killing the tumor cells.
  • viruses are replication competent.
  • they are selectively replication competent in tumor cells. This means that either they replicate in tumor cells and not in non-tumor cells, or that they replicate more effectively in tumor cells than in non-tumor cells.
  • Cells in which the virus is able to replicate are permissive cells.
  • a virus of the invention preferably has a greater ability than an unmodified parent strain to infect or replicate in a tumor cell, to kill tumor cells or to spread between cells in tissues. Preferably this ability is a statistically significant greater ability.
  • a virus according to the invention may have up to 1.1 fold, 1.2 fold, 1.5 fold, 2 fold, 5 fold, 10 fold, 20 fold, 50 fold or 100 fold the capacity of the unmodified parent strain in respect of the property being tested.
  • the viruses of the invention may be modified to carry heterologous nucleic acid sequences.
  • the heterologous nucleic acid encodes a hexosyl transferase.
  • the nucleic acid sequence encodes a galactosyl transferase.
  • the nucleic acid sequence encodes alpha 1 ,3-galactosyltransferase.
  • a1 ,3-galactosyltransferase refers to any enzyme capable of synthesizing alpha-gal epitopes.
  • the enzyme is produced in most mammals with the exception of humans, apes and Old World monkeys.
  • the carbohydrate structure produced by the enzyme is immunogenic in human and most healthy people have high titer natural anti alpha-gal antibodies, also referred to as "anti-Gal" antibodies.
  • alpha 1 ,3GT refers to a mouse alphal ,3GT (e.g., Mus musculus - nucleotides 460 to 1680 of GENBANK Accession No. NM_010283) and its gene product, as well as its functional mammalian counterparts (e.g., other New World monkeys, prosimians and non-primate mammals, but not Old World monkeys, apes and humans).
  • alphal , 3GT refers common marmoset gene (e.g., Callithrix jacchus - GENBANK Accession No.
  • bovine alpha 1 ,3GT e.g., Bos taurus - GENBANK Accession No. NM_17751 1
  • feline alphal , 3GT e.g., Felis catus - GENBANK Accession No. NM_001009308
  • ovine alphal , 3GT e.g., Ovis aries - GENBANK Accession No. NM_001009764
  • rat alphal , 3GT e.g., Rattus norvegicus - GENBANK Accession No. NM_145674
  • porcine alphal ,3GT e.g., Sus scrofa - GENBANK Accession No. NM_213810
  • alphal ,3GT variants include but are not limited to naturally occurring functional mammalian alphal ,3GT variants, as well as non-naturally occurring variants generated by recombinant or other means (e.g., 1 , 2, 3, 4 or 5 amino acid substitutions, deletions, or additions, preferably corresponding to a residue from a functional mammalian alphal , 3GT homolog) are contemplated to find use in the compositions and methods of the present invention.
  • truncated forms of a mammalian alphal ,3GT which retain catalytic activity, are employed (e.g., GGTA1 lacking 90 amino acid N-terminal stem region).
  • GGTA1 lacking 90 amino acid N-terminal stem region.
  • deletion of 3 amino acids from the C-terminus of this enzyme results in its complete inactivation (Henion, T.R., B.A. Macher, F. Anaraki and U. Galili, Glycobiology 4: 193-201 , 1994).
  • Alphal , 3-galactosyltransferase has been previously engineered in adenovirus and used for cellular transfer (Deriy et al. Glycobiology, 12(2), 135 ⁇ 14 2002; Deriy et al. Cancer Gene Therapy, 12(6), 528-39, 2005).
  • Alpha 1 ,3-galactosy transferase has never been engineered to be expressed by oncolytic viruses for the combination lysis/immunization method described herein.
  • the heterologous nucleic acid sequence is preferably operably-linked to a control sequence permitting expression of said gene in a cell in vivo.
  • Viruses of the invention may thus be used to deliver the heterologous nucleic acid sequences to a cell in vivo where it will be expressed.
  • a method of preparing the oncolytic virus as defined herein which comprises the step of incorporating a nucleic acid encoding a hexosyl transferase enzyme into the genome of said oncolytic virus.
  • the heterologous nucleic acid sequence may be inserted into the viral genome by any suitable technique such as homologous recombination of HSV strains with, for example, plasmid vectors carrying the gene flanked by HSV sequences.
  • the heterologous nucleic acid sequence may be introduced into a suitable plasmid vector comprising herpes viral sequences using cloning techniques well-known in the art.
  • said incorporating step comprises cloning.
  • the heterologous nucleic acid sequence may be inserted into the viral genome at any location provided that oncolytic properties are still retained.
  • Heterologous nucleic acid sequence may be inserted at multiple sites within the virus genome. For example, from 2 to 5 genes may be inserted into the genome.
  • the transcribed sequence of the heterologous nucleic acid sequence is preferably operably linked to a control sequence permitting expression of the gene in a tumor cell.
  • the term "operably linked” refers to a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner.
  • a control sequence "operably linked" to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequence.
  • the control sequence comprises a promoter allowing expression of the heterologous nucleic acid sequence and a signal for termination of transcription.
  • the promoter is selected from promoters which are functional in mammalian, preferably human tumor cells.
  • the promoter may be derived from promoter sequences of eukaryotic genes.
  • Viral promoters may also be used, for example the Moloney murine leukemia virus long terminal repeat (MMLV LTR) promoter or other retroviral promoters, the human or mouse cytomegalovirus (CMV) IE promoter, or promoters of herpes virus genes including those driving expression of the latency associated transcripts.
  • MMLV LTR Moloney murine leukemia virus long terminal repeat
  • CMV human or mouse cytomegalovirus
  • herpes virus genes including those driving expression of the latency associated transcripts.
  • Expression cassettes and other suitable constructs comprising the heterologous nucleic acid sequence and control sequences can be made using routine cloning techniques known to persons skilled in the art (see, for example, Sambrook et al., 1989, Molecular Cloning-A laboratory manual; Cold Spring Harbor Press).
  • a virus of the invention may further comprise a heterologous nucleic acid sequence encoding the tet repressor/VP16 transcriptional activator fusion protein under the control of a strong promoter (e.g. the CMV IE promoter) and the heterologous nucleic acid sequence may be under the control of a promoter responsive to the tet repressor VP 16 transcriptional activator fusion protein previously reported (Gossen and Bujard, 1992, Gossen et al, 1995).
  • a strong promoter e.g. the CMV IE promoter
  • expression of the heterologous nucleic acid sequence would depend on the presence or absence of tetracycline.
  • a virus of the invention may comprise two or more heterologous nucleic acid sequences, for example from 2 to 3, 4 or 5 heterologous nucleic acid sequences. More than one gene and associated control sequences could be introduced into a particular oncolytic virus strain either at a single site or at multiple sites in the virus genome. Alternatively, pairs of promoters (the same or different promoters) facing in opposite orientations away from each other, each driving the expression of an heterologous nucleic acid sequence may be used.
  • compositions of the oncolytic viruses of the present invention can be formulated in any conventional manner using one or more physiologically acceptable carriers or excipients.
  • the pharmaceutical compositions of the present invention can be formulated for a variety of modes of administration, including systemic, topical or localized administration. Techniques and formulations can be found in, for example, Remington's Pharmaceutical Sciences, Meade Publishing Co., Easton, Pa.
  • the oncolytic virus composition is formulated for intravenous administration, intramuscular administration, intraperitoneal administration, intratumoral administration, subcutaneous administration, oral administration, rectal administration, intravaginal administration, intranasal administration, transmucosal administration or transdermal administration.
  • the oncolytic virus composition is formulated for intravenous administration, intramuscular administration, intraperitoneal administration,
  • subcutaneous administration oral administration, rectal administration, intravaginal administration, intranasal administration, transmucosal administration or transdermal administration.
  • suitable oncolytic virus to cancerous cells that are to be treated may be performed using naked virus or by encapsulation of the virus in a carrier, e.g. in nanopartides, liposomes or other vesicles.
  • Administration is preferably in a "therapeutically effective amouni", this being sufficient to show benefit to the individual.
  • the actual amount administered, and rate and time- course of administration, will depend on the nature and severity of the tumor being treated. Prescription of treatment, e.g. decisions on dosage etc, is within the
  • the oncolytic virus may be administered at any therapeutically effective dosage amount.
  • Therapeutically effective dosages may be about, but not limited to, 10 3 , about 1 Q 4 , about 10 5 , about 10 6 , about 10 7 , about 10 8 or about 10 9 plaque forming units (pfu).
  • the pharmaceutical compositions of the present invention can be formulated in liquid solutions, preferably in physiologically compatible buffers, such as Hank's solution or Ringer's solution.
  • the pharmaceutical compositions may be formulated in solid form and redissolved or suspended immediately prior to use. Lyophilized forms of the pharmaceutical composition are also suitable.
  • Systemic administration can also be by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, bile salts, and fusidic acid derivatives.
  • detergents may be used to facilitate permeation.
  • Transmucosal administration can occur using nasal sprays or suppositories.
  • the vector particles of the invention can be formulated into ointments, salves, gels, or creams as generally known in the art.
  • a wash solution can also be used locally to treat an injury or inflammation in order to accelerate healing.
  • compositions can be formulated for parenteral administration by injection, e.g. by bolus injection or continuous infusion.
  • Formulations for injection can be presented in a unit dosage form, e.g. in ampoules or in multi-dose containers, with an optionally added preservative.
  • the pharmaceutical compositions can further be formulated as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain other agents including suspending, stabilizing and/or dispersing agents.
  • the invention also includes a kit comprising a pharmaceutical composition of the invention and a delivery device for delivering the composition to a subject.
  • the delivery device may be a squeezable spray bottle, a metered-dose spray bottle, an aerosol spray device, an atomizer, a dry powder delivery device, a self- propelling solvent/powder-dispensing device, a syringe, a needle, a tampon, or a dosage measuring container.
  • the kit may further comprise an instructional material as described herein.
  • viruses of the invention may be used in methods of cancer therapy of the human or animal body.
  • viruses of the invention may be used in the oncolytic treatment of cancer, either with or without additional pro-drug therapy or stimulation of an anti-tumor immune response.
  • Virus compositions of the invention may be used in the therapeutic treatment of any solid tumor in a mammal, preferably in a human.
  • a method of treating an individual with a neoplasm which comprises the steps of:
  • the method is directed to treating an individual with cancer by administering an effective amount of an oncolytic virus composition wherein virus infection causes expression of hexosyl transferase prior to lysis.
  • the oncolytic virus is administered in an effective amount to infect at least one cancer cell in the individual.
  • alpha-gal epitopes are inserted into the infected cell membrane within 10 hours of infection.
  • alpha-gal epitopes insert into the infected cell membrane prior to lysis of the infected cell.
  • the method is directed to treating an individual with cancer.
  • the method is directed to treating an individual with cancer.
  • the modified cell membrane actively targets membrane fragments for antigen presenting cell recruitment.
  • the enzyme is sufficient to express alpha-gal epitopes on the infected cell membrane prior to lysis.
  • alpha1 ,3-galactosyltransferase expression is sufficient to induce an effective immune response to the infected cell after lysis.
  • the alpha-gal epitopes are sufficient to attract antigen- presenting cells to the tumor site.
  • the enzyme is expressed within 4 hours from infection.
  • the alpha-gal epitopes are sufficient to bind anti-gal antibodies in the tumor site.
  • Anti-Gal mediated destruction of tumors described in this method may be achieved by injection of gene therapy vectors containing the alpha1 ,3-galactosyltransferase gene.
  • the present invention contemplates a method of treating melanoma patients with multiple metastases comprising intratumoral injections of replication competent oncolytic virus containing the alphal ,3-galactosyltransferase gene.
  • intratumoral injection of viral compositions results in transduced tumor cells expressing alpha-gal epitopes, wherein these tumor cells induce intratumoral inflammation prior to lysis. Intratumoral inflammation recruits antigen presenting cells such as dendritic cells, macrophages and certain B-cells.
  • Infected cells can be either lysed by the engineered replication competent virus compositions described herein, or alternatively destroyed by the natural anti-Gal antibody bound to alpha-gal epitopes on the transduced cells via the complement dependent cytolysis (CDC) or antibody dependent cell mediated cytolysis (ADCC) mechanisms.
  • CDC complement dependent cytolysis
  • ADCC antibody dependent cell mediated cytolysis
  • the present invention contemplates a method of treating colorectal carcinoma patients having multiple metastases in the colon and in the liver comprising intratumoral injections of oncolytic virus containing the alpha 1 ,3- galactosyltransferase gene.
  • the injection comprises colonoscopy or laparoscopy as means of delivering the viral vector into the tumor lesions.
  • the present invention contemplates a method of treating lung carcinoma patients having multiple metastases in the lungs comprising intratumoral injections of oncolytic virus containing the alpha 1 ,3-galactosyltransferase gene.
  • the injection comprises bronchoscopy.
  • the present invention contemplates a method of treating patients with urinary bladder carcinoma comprising an oncolytic virus containing the alpha 1 ,3-galactosy transferase gene viral vector.
  • the vector is administered by means of cystoscopy.
  • the present invention contemplates a method of treating patients with pancreatic adenocarcinoma comprising an oncolytic virus containing the alpha 1 ,3-galactosy transferase gene viral vector.
  • the vector is administered by means of endoscopy or laparoscopy.
  • the present invention contemplates a method of treating patients with mammary carcinoma comprising an oncolytic virus containing the alpha 1 ,3-galactosyltransferase gene viral vector.
  • the vector is administered by direct injection into the tumor.
  • Alternative methods to deliver the alpha 1 ,3-galactosyltransferase gene can be performed also with any type of viral and non-viral vector which can deliver genes.
  • these methods include, but are not limited to adenovirus vector, adenovirus helper virus, retrovirus vector, lentivirus vector, naked DNA vectors, herpes virus or naked RNA vectors or DNA vectors.
  • the present invention contemplates a method to administer vectors containing the alpha 1 ,3-galactosyltransferase gene by injection into melanoma lesions or any other tumor lesion, whereby the epitopes insert into the tumor cell membranes.
  • the anti-Gal IgG will bind to the tumor cell membranes expressing alpha-gal epitopes and will target them to antigen presenting cells for eliciting a systemic immune response also against non-treated tumor lesions that express the tumor antigens.
  • the present invention contemplates generation of adaptive anti-tumor immunity, driven by targeting by the anti-Gal antibody to antigen presenting cells by any composition introduced into the tumor mass that results in the in situ binding of this natural antibody to the tumor cells.
  • the viruses of the invention may be used in a patient, preferably a human patient, in need of treatment.
  • a patient in need of treatment is an individual suffering from cancer, preferably an individual with a solid tumor.
  • the aim of therapeutic treatment is to improve the condition of a patient.
  • therapeutic treatment using a virus of the invention alleviates the symptoms of the cancer.
  • a method of treating cancer comprising administering a therapeutically effective amount of a virus of the invention to a patient suffering from cancer or having a neoplasm or tumor in need of treatment.
  • an oncolytic virus as defined herein or a pharmaceutical composition as defined herein for use in the treatment of cancer is provided.
  • an oncolytic virus of the invention administered to an individual suffering from a tumor will typically kill the cells of the tumor thus decreasing the size of the tumor and/or preventing spread of malignant cells from the tumor while also recruiting antigen presenting cells (APCs) to the tumor site and inducing a protective anti-tumor immune response.
  • APCs antigen presenting cells
  • One method of administering therapy involves combining the virus with a
  • Suitable carriers and diluents include isotonic saline solutions, for example phosphate- buffered saline.
  • the compositions described herein can be administered using conventional administration routes to promote viral infection at a tumor site.
  • the compositions are administered via injection (for example intraperitoneal, intramuscular, intravenous, subcutaneous), inhalation or insulation (for example either through the mouth or the nose transmucosally or intranasally) or by oral, buccal, parenteral or rectal administration routes.
  • the composition is administered by direct injection into target tissue which may be the tumor or a blood vessel supplying the tumor.
  • the amount of virus administered is in the case of HSV in the range of from 10 4 to 10 10 plaque forming units (pfu), preferably from 10 5 to 10 8 pfu, more preferably about 10 6 to 10 8 pfu.
  • pfu plaque forming units
  • up to 500 ⁇ 1 typically from 1 to 200 ⁇ 1 preferably from 1 to 10 ⁇ 1 of a pharmaceutical composition of the virus and a pharmaceutically acceptable suitable carrier or diluent would be used for injection.
  • larger volumes up to, but not limited to 10 ml may also be used, depending on the tumor and the inoculation site.
  • the routes of administration and dosages described are intended only as a guide since a skilled practitioner will be able to determine readily the optimum route of administration and dosage.
  • the dosage may be determined according to various parameters, especially according to the location of the tumor, the size of the tumor, the age, weight and condition of the patient to be treated and the route of administration.
  • the dosage and dosage frequency may first be optimized pre-clinically by studying the properties of the virus in tissue culture and in a suitable animal model.
  • the virus may be combined with a pharmaceutically acceptable carrier or diluent to produce a pharmaceutical composition.
  • a suitable carrier for most viruses would be a buffered saline solution, such as phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • the pharmaceutical composition will ideally be formulated to enable administration of the appropriate dose of virus in a volume of up to 1 ml, although there may be cases where up to a 10 ml volume is administered. In cases where individual tumor lesion volumes are deemed too small to be treated with the optimal dose, it is possible that the optimal dose is administered by treatment of multiple lesions, each administered with a fraction of the total optimal dose. It is also envisaged that multiple doses may be required to elicit an effective anti-tumor response.
  • the dose interval may be determined based on efficacy data obtained in preclinical models or from clinical experience obtained with a similar virus strain.
  • the virus is administered by direct injection into the tumor.
  • the virus may also be administered systemically or by injection into a blood vessel supplying the tumor.
  • the virus may also be administered as an intravesical treatment; such as might be used for treatment of cancers of the bladder.
  • the optimum route of administration will depend on the location and size of the tumor.
  • the method is a method of treating an individual having a neoplasm comprising administering an effective amount of a modified oncolytic virus comprising a nucleic acid encoding a galactosyl transferase enzyme.
  • the oncolytic viruses of the present invention may be formulated for administration by, for example, injection, inhalation or insulation (either through the mouth or the nose) or by oral, buccal, parenteral or rectal administration.
  • the oncolytic viruses useful in the present methods may be administered to a subject with prostate, breast, lung, liver, endometrial, bladder, colon or cervical carcinoma; adenocarcinoma; melanoma; lymphoma; glioma; or sarcomas such as soft tissue and bone sarcomas.
  • the invention is directed to the engineered oncolytic virus of the invention for the treatment or prevention of cancer, including, but not limited to, neoplasms, tumors, metastases, or any disease or disorder characterized by
  • the cancer can be a multifocal tumor.
  • types of cancer and proliferative disorders to be treated with the therapeutics of the invention include, but are not limited to, leukemia (e.g. myeloblasts, promyelocytic, myelomonocytic, monocytic, erythroleukemia, chronic myelocytic (granulocytic) leukemia, and chronic lymphocytic leukemia), lymphoma (e.g.
  • fibrosarcoma myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, angiosarcoma, endotheliosarcoma, Ewing's tumor, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, renal cell carcinoma, hepatoma, Wilms' tumor, cervical cancer, uterine cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, oligodendroglioma, melanoma, neuroblastoma, retinoblastoma, dysplasia and hyperplasia.
  • therapeutic compounds of the invention are administered to patients having prostate cancer (e.g., prostatitis, benign prostatic hypertrophy, benign prostatic hyperplasia (BPH), prostatic paraganglioma, prostate adenocarcinoma, prostatic intraepithelial neoplasia, prostato- rectal fistulas, and atypical prostatic stromal lesions).
  • prostate cancer e.g., prostatitis, benign prostatic hypertrophy, benign prostatic hyperplasia (BPH), prostatic paraganglioma, prostate adenocarcinoma, prostatic intraepithelial neoplasia, prostato- rectal fistulas, and atypical prostatic stromal lesions.
  • the medicaments of the present invention are used for the treatment of cancer, glioma, liver carcinoma and/or colon carcinoma.
  • the treatment and/or prevention of cancer includes, but is not limited to, alleviating symptoms associated with cancer, the inhibition of the progression of cancer,
  • neoplasm refers to an abnormal growth of tissue.
  • a neoplasm may be benign or malignant.
  • a malignant neoplasm is referred to as a cancer.
  • Cancers differ from benign neoplasms in the ability of malignant cells to invade other tissues, either by direct growth into adjacent tissue through invasion or by implantation into distant sites by metastasis (i.e., transport through the blood or lymphatic system).
  • metastasis i.e., transport through the blood or lymphatic system.
  • the methods of the present invention are suitable for the treatment of benign and malignant neoplasms (cancer).
  • a superficial neoplasm is one located on the outer surface of the body that has confined itself and not spread to surrounding tissues or other parts of the body.
  • An internal neoplasms located on an internal organ or other internal part of the body.
  • An invasive neoplasm is a neoplasm that has started to break through normal tissue barriers and invade surrounding areas, e.g., an invasive breast cancer that has spread beyond the ducts and lobules.
  • neoplasms contemplated for treatment by the method disclosed herein includes the following categories: (a) abdominal neoplasms including peritoneal neoplasms and retroperitoneal neoplasms; (b) bone neoplasms including femoral neoplasms, skull neoplasms, jaw neoplasms, manibular neoplasms, maxillary neoplasms, palatal neoplasms, nose neoplasms, orbital neoplasms, skull base neoplasms, and spinal neoplasms; c) breast neoplasms including male breast neoplasms, breast ductal carcinoma, and phyllodes tumor; (d) digestive system neoplasms including biliary tract neoplasms, bile duct neoplasms, common bile duct neoplasms, gall bladder n
  • endocrine gland neoplasms including adrenal gland neoplasms, adrenal cortex neoplasms, adrenocortical adenoma, adrenocortical carcinoma, multiple endocrine neoplasia, multiple endocrine neoplasia type 1 , multiple endocrine neoplasia type 2a, multiple endocrine neoplasia type 2b, ovarian neoplasms, granulosa cell tumor, luteoma, Meigs' Syndrome, ovarian Sertoli-Leydig cell tumor, thecoma, pancreatic neoplasms, paraneoplastic endocrine syndromes, parathyroid neoplasms, pituitary neoplasms, Nelson Syndrome, testicular
  • compositions of the present invention can be administered alone or in combination with other types of cancer treatment strategies (e.g., radiation therapy, chemotherapy, hormonal therapy, immunotherapy and anti-tumor agents).
  • anti-tumor agents include, but are not limited to, cisplatin, ifosfamide, paclitaxel, taxanes, topoisomerase I inhibitors (e. g., CPT-1 1 , topotecan, 9-AC, and GG-21 1), gemcitabine, vinorelbine, oxaliplatin, 5-fluorouracil (5-FU), leucovorin, vinorelbine, temodal, and taxo.
  • the pAd shuttle plasmid containing the alpha 1 ,3GT cDNA that allows for homologous recombination of the cDNA into the replication defective adenovirus vector.
  • the a1 ,3GT gene was inserted downstream of the cytomegalovirus (CMV) promoter which is a very effective promoter in mammalian cells.
  • CMV cytomegalovirus
  • the generated adenovirus vector containing the inserted mouse a1 ,3GT gene under the CMV promoter was propagated in the human kidney cell line 293 (ATCC) which contains the E1 complementing viral gene.
  • the viral clone isolated was assayed for presence of a1 ,3GT cDNA that produces catalytically active enzyme, by transduction of 293 cells and analysis of alpha- gal epitope expression after 24 h by flow cytometry following Bandeiraea simplicifolia IB4 (BS lectin) binding.
  • the BS lectin binds specifically to alpha-gal epitopes.
  • AdaGT The replication defective adenovirus contacting the a1 ,3GT gene was designated AdaGT and was further propagated in the 293 cell line which contains the E1 complementing viral gene.
  • concentration of AdaGT was determined as multiplicity of infection (MOI) units and defined as the highest dilution of adenovirus vector that displayed cytopathic effects in 293 cells, 6 days post infection.
  • MOI multiplicity of infection
  • the ability of AdaGT to induce synthesis and expression of alpha - gal epitopes on human tumor cells was studied with the human cervical carcinoma HeLa cell line, which like other malignant or normal human cells, it lacks alpha-gal epitopes.
  • a1 ,3GT mRNA originating from AdaGT was determined by reverse transcriptase polymerase chain reaction (RT-PCR) using upstream and downstream primers specific to the mouse a1 ,3GT gene (5 '- ATG AATGTCAAG G G AAAAG-3 ' (SEQ ID NO: 1) and 3'-TCAGACATTATTTCTAACCA-5' (SEQ ID NO: 2)). Based on this analysis a1 ,3GT mRNA was found in the cytoplasm of the HeLa cells, 4h after transduction. The actual appearance of the a1 ,3GT enzyme in the cytoplasm was determined by the catalytic activity of this enzyme.
  • a1 ,3GT transfers galactose from the sugar donor UDP-Gal to nine terminal N- acetyllactosamine residues on N-linked carbohydrate chains of the protein asialofetuin coating ELISA wells. De novo synthesized alpha-gal epitopes were identified by ELISA with the monoclonal anti-Gal antibody M86 (Galili et al. Transplantation, 65:1 129-1 132, 1998). By using this assay, catalytic activity of a1 ,3GT was first detected in the transduced HeLa cells after 6 h (see timeline in Figure 3).
  • AdaGT AdaGT is replication defective and cannot proliferate in dividing cells
  • each cell division of the transduced HeLa cells results in the decrease in the number of a1 ,3GT copies by 50%. Accordingly, expression of alpha-gal epitopes decreases on the surface of the cells so that within 2 weeks post transduction no alpha-gal epitopes are detected on the cells and no a1 ,3GT gene is detected within the cells.
  • Example 2 Expression of alpha-gal epitopes on B16-BL6 melanoma cells
  • alpha 1 ,3-galactosyltransferase gene was inserted into a replication incompetent adenovirus vector as previously described (Deriy et al. Glycobiology 2002, 12: 135).
  • the resulting vector is designated AdaGT and is very effective in inducing expression of alpha-gal epitopes on human tumor cells (Deriy L et al, supra).
  • the expression of alpha-gal epitopes was determined on AdaGT transduced B16-BL6 melanoma cells. These cells are a subclone of B16 melanoma and are referred to as BL6 cells.
  • the transduction of BL6 cells with AdaGT results in intracellular production of alpha 1 ,3-galactosyltransferase that is encoded by the alpha 1 ,3-galactosyltransferase gene within the transducing AdaGT.
  • the de novo expression of alpha-gal epitopes on cell surface glycoconjugates following synthesis of this epitope by the alpha 1 ,3-galactosyltransferase was evaluated 48 hour post
  • Alpha-gal epitopes were detected by the binding of Bandeiraea (Griffonia) simplicifolia IB4 lectin (BS lectin - a lectin specific to a-gal epitopes) as measured by flow cytometry and by the binding of the anti-Gal antibody to the transduced cells as measured by ELISA.
  • Bandeiraea Griffonia
  • simplicifolia IB4 lectin BS lectin - a lectin specific to a-gal epitopes
  • BL6 cells transduced with AdaGT displayed a significant shift following BS lectin binding, as measured by flow cytometry, in comparison to B16 cells transduced with the control "empty" adenovirus that lacks the alpha1 ,3-galactosyltransferase gene insert (referred to as BL6 A dcont cells) ( Figure 4A).
  • BL6 A dcont cells the control "empty" adenovirus that lacks the alpha1 ,3-galactosyltransferase gene insert
  • Example 3 Efficacy of BL6 A daGT cells in eliciting a protective anti-tumor immune response
  • AdaGT transduced tumor cells were studied in alpha 1 ,3- galactosyltransferase knockout mice, as detailed in Deriy et al. Cancer Gene Therapy 12: 528-539, 2005).
  • the B16-BL6 cells served as the tumor model.
  • Anti-Gal producing mice were vaccinated with 2x10 6 irradiated BL6 AdccG T cells, or with 2x10 6 irradiated BL6 cells that were transduced with the control parental adenovirus vector lacking the alpha 1 ,3-galactosy transferase gene.
  • BL6 AdC0 nt The latter cells, referred to as BL6 AdC0 nt, did not express alpha-gal epitopes and served for immunization of the control mice.
  • the immunization was repeated after one week.
  • the mice were challenged subcutaneously with 0.2x10 6 or 0.5x10 6 live BL6 cells. Tumor development was monitored for 2 months.
  • Two thirds of the mice immunized with BL6 Adtt GT cells and challenged with 0.2x10 6 non-transduced parental BL6 cells were protected against the challenge, whereas only 20% of the control group did not develop tumor (Figure 5A).
  • Example 4 Kinetics of alpha-gal epitope expression vs. cytolysis in tumor cells infected with Herpes Virus containing the a1,3GT gene (HSVCGT)
  • the objective of this experiment is to determine in vitro the timeline for the expression of alpha-gal epitopes on human and mouse tumor cells following the infection of the cells with oncolytic herpes virus containing the a1 ,3GT gene (HSVXGT). This analysis is required in order to confirm that the expression of alpha-gal epitopes on infected cells occurs prior to the cytolysis of the cells by this oncolytic virus. It is assumed that once the alpha-gal epitopes are expressed on cell membranes they will bind the natural anti- Gal antibody even after the cells are lysed by the oncolytic virus HSV XG T.
  • the immune complexes formed between anti-Gal and fragmented tumor cell membranes (because of the cell lysis) will be effectively targeted for uptake by APCs such as dendritic cells and macrophages, as a result of the interaction between the Fc portion of the
  • HSV a G T the a1 ,3GT gene under the CMV promoter is amplified by PCR from the replication defective virus AdaGT described above and in (Deriy et al 2002, supra). The gene under CMV promoter is inserted into an oncolytic HSV and suspensions of the resulting HSV a G T are prepared by propagation in HeLa cells according to methods known to those skilled in the art.
  • alpha-gal epitopes is determined on HSV a G T infected B16F10 (B16) mouse melanoma cells and on human HeLa cervical carcinoma cells.
  • B16 B16F10
  • the study with the mouse melanoma cells is required to determine the subsequent feasibility of the in vivo study with these cells in mice.
  • the B16 melanoma is the only known mouse tumor cell line that lacks alpha-gal epitopes and thus it simulates human tumor cells in the lack of this carbohydrate epitope.
  • the study with the HeLa cells is required to confirm that the assumption that alpha-gal epitope expression in HSV tt GT infected human tumor cells occurs prior to the cytolysis of the cells by this virus.
  • a suspension of HSV tt GT virus is added to monolayers of B16 cells at a concentration of 1x10 6 -2x10 7 pfu (plaque forming units)/ml.
  • the infected cells are detached every 2 hours and the expression of the a1 ,3GT gene of the virus is determined by the appearance of a1 ,3GT mRNA.
  • mRNA is detected by reverse transcriptase polymerase chain reaction (RT-PCR) using upstream and downstream primers specific to the mouse a1 ,3GT gene (5'-ATGAATGTCAAGGGAAAAG-3' (SEQ ID NO: 1) and 3' -TCAGACATTATTTCTAACCA-5' (SEQ ID NO: 2)) as described for AdaGT above.
  • RT-PCR reverse transcriptase polymerase chain reaction
  • the actual appearance of the a1 ,3GT enzyme in the cytoplasm of the infected cells is determined by the catalytic activity of this enzyme in ELISA using asialofetuin as an acceptor for the enzyme and UDP-Gal is used as the sugar donor.
  • Asialofetuin further functions as a solid phase antigen coating ELISA wells.
  • alpha-gal epitopes are identified by ELISA with the monoclonal anti-Gal antibody M86 (Galili, 1998, supra).
  • M86 monoclonal anti-Gal antibody
  • the subsequent appearance of alpha-gal epitopes is detected by the binding of Bandeiraea (Griffonia) simplicifolia IB4 lectin (BS lectin - a lectin specific to alpha-gal epitopes) as measured by flow cytometry and by the binding of the anti-Gal antibody to the infected cells processed to coat ELISA by drying the cells in ELISA wells.
  • Bandeiraea Griffonia
  • simplicifolia IB4 lectin BS lectin - a lectin specific to alpha-gal epitopes
  • the alpha-gal epitope expression is further quantified by the sensitive monoclonal anti- Gal M86 antibody binding assay called ELISA Inhibition Assay which measures the number of alpha-gal epitopes per cell (Galili et a/.1998, supra).
  • ELISA Inhibition Assay which measures the number of alpha-gal epitopes per cell (Galili et a/.1998, supra).
  • An "empty" HSV i.e. virus lacking the a1 ,3GT gene
  • the cell cultures continue to be monitored microscopically for cytolysis in order to determine the period post infection in which the cells undergo cytolysis.
  • Example 5 Efficacy of HSV aG T as an in situ oncolytic virus
  • HSV tt GT as an oncolytic virus and as means to generate protective antitumor immunity against various tumor associated antigens (TAA) is determined according to the studies (Galili et al. J Immunol.; 178: 4676-87, 2007; Abdel-Motal et al. Cancer Immunol lmmunother.;58: 1545-55, 2009.) These studies are performed in alpha 1 ,3-galactosyltransferase knockout mice producing the anti-Gal antibody.
  • the B16 cells serve as the tumor model.
  • Anti-Gal producing alpha1 ,3-galactosyltransferase knockout mice are injected subcutaneously in the right abdominal flank with 1x10 6 B16 cells.
  • the B16 cells develop into a melanoma tumor lesion with a diameter of 4-5 mm within 5-6 days.
  • HSV XG T at an amount ranging between 10 3 - 10 6 pfu in a volume of 0.1 ml is injected into the tumors.
  • Oncolytic HSV lacking the inserted a1 ,3GT gene (“empty" HSV) is used as control virus incapable of inducing the expression of a-gal epitopes. The mice are monitored for 2 months for the
  • HSVXGT which is twice the amount displaying complete elimination of the tumor (i.e. 100% oncolysis) is determined and used for further studies evaluating the protective anti-tumor immune response elicited by this treatment.
  • Comparison with the control HSV lacking the a1 ,3GT gene determines whether HSV XG T is more, or less effective than the control virus in inducing oncolysis.
  • Example 6 Efficacy of HSV aG T infected lesions in eliciting a protective anti-tumor immune response against distant metastases
  • Anti-Gal producing alpha 1 ,3-galactosyltransferase knockout mice are injected subcutaneously in the right abdominal flank with 1x10 6 B16 cells and subcutaneously in the left flank with 1x10 4 , 1x10 5 or 1x10 6 B16 cells.
  • the tumor developing in the left flank represents a distant metastasis which may not develop into a tumor lesion if the injection of HSV a G T into the right flank lesion converts it into a vaccine that elicits a systemic protective anti-tumor response.
  • the right flank tumor When the right flank tumor reaches the size (diameter) of 4-5 mm (within 5-6 days) it is injected with 0.1 ml suspension HSVaGT at an amount that is twice the amount that induces 100% cytolysis of the melanoma lesion.
  • Oncolytic HSV lacking the inserted a1 ,3GT gene is used as control virus incapable of inducing the expression of alpha-gal epitopes.
  • the growth of the tumor in the left flank is monitored for 2 months. Conversion of the tumor injected with HSV a G T into a vaccine results in prevention of the distant metastases in the left flank from growing into a detectable lesion when the left flank is injected with 1x10 4 , 1x10 5 or 1x10 6 B16 cells.
  • Anti-Gal producing alpha 1 ,3-galactosyltransferase knockout mice are injected subcutaneously in the right abdominal flank with 1x10 6 B16 cells.
  • the tumor reaches the size (diameter) of 4-5 mm (within 5-6 days) it is injected with 0.1 ml suspension HSV tt GT at an amount that is twice the amount that induces 100% cytolysis of the melanoma lesion.
  • Oncolytic HSV lacking the inserted a1 ,3GT gene is used as control virus incapable of inducing the expression of alpha -gal epitopes.
  • the left flank is injected with 0.3x10 6 , 1x10 6 , 3x10 6 , or 10x10 6 B16 cells as tumor challenge.
  • the induction of a protective anti-tumor immune response as a result of injection of HSV tt GT into the right flank tumor results in prevention of the challenging tumor cells in the left flank from developing into a tumor lesion.
  • the prevention of tumor growth is more effective (i.e. prevention of higher number of challenging tumor cells from developing into a lesion) when the tumor in the right flank is injected with HSV tt GT than with HSV lacking the a1 ,3GT gene.
  • Example 8 Production and efficacy of a replication competent oncolytic adenovirus that encodes a functional alpha 1,3-galactosyltransferase gene
  • CRAd-aGT Ad5/3-A24-aGT CRAd
  • E3 gene was replaced with the murine alpha 1 ,3-galactosyltransferase (a1 ,3GT) gene.
  • a1 ,3GT synthesizes the carbohydrate antigen galactose-alpha-1 ,3- galactosyl-beta-1 ,4-N-acetyl-glucosamine-R (alpha-gal).
  • alpha-gal carbohydrate antigen galactose-alpha-1 ,3- galactosyl-beta-1 ,4-N-acetyl-glucosamine-R
  • A549 (human lung carcinoma) and A375 (human melanoma) cell lines were purchased from the European Collection of Authenticated Cell Cultures (ECCAC).
  • A549 cells were maintained in F-12K + 10% fetal bovine serum (FBS).
  • A375 cells were maintained in Dulbecco's Modified Eagle Medium (DMEM) + 10% fetal bovine serum. Both cell lines were incubated at 37°C and 5% C0 2 .
  • Virus production was performed under contract by Imanis Life Sciences (Rochester, Minneapolis, US). Virus construction was performed using established shuttle and large adenoviral plasmids and recombinant DNA methods exactly as described in detail (Danthinne and Werth, 2000; Danthinne, 2001).
  • the 1.1 Kb murine a1 ,3GT gene (GenBank accession number M85153) was synthesized by Integrated DNA Technologies (IDT) and cloned into an Ad5 E3 shuttle vector with a
  • the CMV promotor 5' of the a1 ,3GT sequence In a cosmid encoding the Ad5 genome, with the fiber knob sequence replaced with that of Ad3, the 24-bp sequence encoding the LTCHEAGF (SEQ ID NO: 3) amino acid sequence was deleted from region E1 a.
  • the E3 sequence was deleted from the Ad5/3 cosmid and replaced with the CMV-aGT sequence, subcloned from the shuttle vector.
  • the green fluorescent protein (GFP) coding sequence was synthesized by IDT and cloned into an Ad5 E1 shuttle vector with a CMV promotor 5' of the GFP sequence.
  • the E1 sequence was deleted from the Ad5/3 cosmid and replaced with the CMV-GFP sequence, subcloned from the shuttle vector.
  • the E3 region was deleted.
  • the chimeric virus structures are shown in Figure 6.
  • HEK 293 cells were transfected with the chimeric virus cosmids and, after 48 hours incubation, crude lysates prepared. For virus amplification, HEK 293 cells were seeded in 15 cm dishes and incubated overnight at 37°C, 5% C0 2 . The next day, the cells were infected with the crude lysate from virus-infected cells and incubated for 48 hours at 37°C, 5% C0 2 , after which time the cells and supernatant were harvested.
  • Virus particles were released from infected cells by three freeze-thaw cycles and recovered from the cell supernatant by ammonium sulphate precipitation (Schagen et a/., 2000). Virus purification was performed by cesium chloride (CsCI) density gradient centrifugation. Briefly, the crude virus preparation was loaded onto a two-step CsCI gradient in a SW28 Beckman centrifuge tube and centrifuged for 2 hours at 20,000 rpm. The virus band was collected and loaded on to a continuous CsCI gradient and centrifuged again.
  • CsCI cesium chloride
  • the final virus band was collected and immediately dialyzed against 4 x 0.5 L GTS buffer (2.5% glycerol, 25 mM NaCI, 20 mM Tris-HCI pH 8.0) for 18 hours at 4°C. Approximately 1 ml dialysed virus suspension was collected and filtered through a 0.22 ⁇ Supor membrane (Pall) then frozen at -70°C. Virus concentration was determined using the OD260-SDS method and the extinction coefficient of 1.1 x 10 12 virus particles (VP) per absorbance 260 unit in the presence of SDS. The virus concentrations were found to be 3.6x10 12 VP/ml for CRAd-aGT and 2.9x10 12 VP/ml for CRAd-GFP. The purity of the virus preparations was determined by A260/A280 and A320/A260 ratios and was found to be within the range associated with pure virus (1.2 to 1.4 and 0.22 to 0.27 respectively) for both viruses.
  • the virus titer was determined by endpoint dilution assay (TCID50) using the well- established Reed-Muench method (Reed and Muench, 1938). Briefly, HEK293 cells were seeded into 24-well plates and incubated overnight at 37°C. The next day, a 10- fold dilution series of CsCI-purified virus stock was added to the seeded HEK293 cells and the cells incubated at 37°C, 5% C0 2 for 14 days. Wells were scored for the presence of cytopathic effects (CPE) by visual assessment under the microscope and the TCID50 calculated using the Reed-Muench equation (Reed and Muench, 1938). The virus titer was determined to be 6.8x10 11 TCI D50/mL for CRAd-aGT and 2.7x10 11 TCID50/ml for the CRAd-GFP.
  • CPE cytopathic effects
  • Sub-confluent monolayers of A549 and A375 cells were harvested from tissue culture flasks using cell dissociation solution (CDS, Sigma-Aldrich). The cells were counted using a hemocytometer and live cells distinguished from dead by Trypan Blue exclusion. The cells were diluted in culture medium and added to sterile 96-well white, tissue culture plates at 90 ⁇ /well to give either 5x10 3 or 1x10 4 cells/well.
  • CDS cell dissociation solution
  • Each virus was diluted 1 :3 in phosphate buffered saline (PBS) + 10% FBS to provide working stock solutions of 1.2x10 12 VP/ml for CRAd-aGT and 9.67x10 11 VP/ml for CRAd- GFP.
  • the working stocks were serially diluted 1 in 5 in PBS + 10% FBS to generate a titration series of 1.2x10 12 - 1.2x10 5 VP/ml for CRAd-aGT and 9.67x10 11 - 9.9x10 4 for CRAd-GFP.
  • the titration series were diluted 1 :10 into the assay plates (10 ⁇ per well of virus to the 90 ⁇ seeded cells) and the plates incubated for 72 hours at 37°C/5% C0 2 .
  • Cell viability was determined using Cell Titre Glo luminescent cell viability reagent (Promega), according to manufacturer's instructions, and the plates read on an EnVision 2102 plate reader (Perkin Elmer). Raw luminescence units (RLU) were plotted against log virus particles/ml.
  • A549 and A375 cells were harvested from tissue culture flasks as described above. After counting, the cells were diluted in culture medium and added to 24-well, tissue culture plates in a volume of 540 ⁇ /well, which equated to 3x10 4 cells/well.
  • Each virus was diluted 1 :10,000 in PBS + 10% FBS (1 :100 and then 1 :100) to provide solutions of 3.6x10 8 VP/ml for CRAd-aGT and 2.9x10 8 for CRAd-GFP. These solutions were diluted 1 :25 twice in PBS + 10% FBS to give virus concentrations of 1.44x10 7 and 5.8x10 5 VP/ml for CRAd-aGT and 1.16x10 7 and 4.6x10 5 VP/ml for CRAd-GFP. The three virus dilutions were diluted a further 1 :10 onto the seeded cells (60 ⁇ virus to the 540 ⁇ cells per well).
  • the cells were washed again in PBS + 0.5% BSA and then incubated with allophycocyanin (APC)-conjugated anti-human IgG (Biolegend), diluted in PBS + 0.5% BSA at 4°C for 30 minutes in the dark.
  • Cells were washed with PBS + 0.5% BSA and analyzed on a Beckman Coulter FC500 flow cytometer. Unstained cells were used to set the voltages for forward scatter, side scatter and fluorescent channels 1 and 4. GFP (channel 1) and APC (channel 4) signals were recorded for test samples. Single color controls were used to set the compensation.
  • the cytolytic effects of CRAd-aGT and control CRAd-GFP on A549 and A375 cells were quantified using a plate-based cell viability assay (Figure 7).
  • the reduction in cell viability observed in wells where cells were infected with either CRAd-aGT or CRAd-GFP was dependent on the concentration of virus.
  • the number of CRAd-aGT virus particles required to reduce cell viability by 50% was lower than for the control CRAd-GFP virus. This is in line with the virus titer measured by TCID50 assay (see Materials and Methods section above).
  • Antigen presenting cells APCs
  • Fey receptors engulf tumor cell debris and dead cells complexed with anti-Gal IgG via activating Fey receptors.
  • Activation of Fey receptors on APCs leads to APC activation, maturation and enhanced presentation of antigen to T cells (Regnault et al., 1999; Rafiq et al., 2002; Platzer et a/., 2014).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Immunology (AREA)
  • Mycology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oncology (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Hematology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Dermatology (AREA)
  • Reproductive Health (AREA)
  • Urology & Nephrology (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)
  • Otolaryngology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
PCT/GB2016/050973 2015-04-07 2016-04-07 Therapeutic compositions and methods of use for treating cancer WO2016162675A1 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
MX2017012867A MX2017012867A (es) 2015-04-07 2016-04-07 Composiciones terapeuticas y metodos de uso para tratar el cancer.
RU2017134519A RU2720984C2 (ru) 2015-04-07 2016-04-07 Терапевтические композиции и способы применения для лечения рака
EP16716896.2A EP3280798B1 (en) 2015-04-07 2016-04-07 Therapeutic compositions of use for treating cancer
CA2981925A CA2981925A1 (en) 2015-04-07 2016-04-07 Therapeutic compositions and methods of use for treating cancer
JP2017552992A JP2018512150A (ja) 2015-04-07 2016-04-07 癌を治療するための治療用組成物及び使用方法
BR112017021700-7A BR112017021700A2 (pt) 2015-04-07 2016-04-07 composições terapêuticas e métodos de uso para o tratamento do câncer
ES16716896T ES2743952T3 (es) 2015-04-07 2016-04-07 Composiciones terapéuticas de uso para el tratamiento del cáncer
KR1020177027601A KR102544032B1 (ko) 2015-04-07 2016-04-07 암을 치료하기 위한 치료 조성물 및 사용 방법
US15/564,774 US20180104288A1 (en) 2015-04-07 2016-04-07 Therapeutic compositions and methods of use for treating cancer
CN201680020049.0A CN107787364A (zh) 2015-04-07 2016-04-07 用于治疗癌症的治疗组合物及使用方法
IL254842A IL254842B (en) 2015-04-07 2017-10-02 Therapeutic preparations and methods for use in cancer treatment
HK18110254.0A HK1251009B (zh) 2015-04-07 2018-08-10 用於治療癌症的治療組合物
US16/698,682 US20200155626A1 (en) 2015-04-07 2019-11-27 Therapeutic compositions and methods of use for treating cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB1505860.5 2015-04-07
GBGB1505860.5A GB201505860D0 (en) 2015-04-07 2015-04-07 Therapeutic compositions and methods of use for treating cancer

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/564,774 A-371-Of-International US20180104288A1 (en) 2015-04-07 2016-04-07 Therapeutic compositions and methods of use for treating cancer
US16/698,682 Division US20200155626A1 (en) 2015-04-07 2019-11-27 Therapeutic compositions and methods of use for treating cancer

Publications (1)

Publication Number Publication Date
WO2016162675A1 true WO2016162675A1 (en) 2016-10-13

Family

ID=53190234

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2016/050973 WO2016162675A1 (en) 2015-04-07 2016-04-07 Therapeutic compositions and methods of use for treating cancer

Country Status (14)

Country Link
US (2) US20180104288A1 (ja)
EP (1) EP3280798B1 (ja)
JP (2) JP2018512150A (ja)
KR (1) KR102544032B1 (ja)
CN (1) CN107787364A (ja)
BR (1) BR112017021700A2 (ja)
CA (1) CA2981925A1 (ja)
ES (1) ES2743952T3 (ja)
GB (1) GB201505860D0 (ja)
HK (1) HK1251009B (ja)
IL (1) IL254842B (ja)
MX (1) MX2017012867A (ja)
RU (1) RU2720984C2 (ja)
WO (1) WO2016162675A1 (ja)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4087920A4 (en) * 2020-01-10 2024-02-28 Univ Hong Kong RECOMBINANT VIRUSES EXPRESSING ALPHA-1, 3-GALACTOSYLTRANSFERASE AND THEIR USES

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2971008B1 (en) 2013-03-14 2018-07-25 Salk Institute for Biological Studies Oncolytic adenovirus compositions
WO2017147265A1 (en) 2016-02-23 2017-08-31 Salk Institute For Biological Studies High throughput assay for measuring adenovirus replication kinetics
KR102471633B1 (ko) 2016-02-23 2022-11-25 솔크 인스티튜트 포 바이올로지칼 스터디즈 바이러스 동역학에 미치는 영향 최소화를 위한 치료용 아데노바이러스의 외인성 유전자 발현
CN110062630A (zh) 2016-12-12 2019-07-26 萨克生物研究学院 肿瘤靶向合成腺病毒及其用途
CN110856724B (zh) * 2018-08-24 2022-05-27 杭州康万达医药科技有限公司 包含核酸及car修饰的免疫细胞的治疗剂及其应用
JPWO2020230838A1 (ja) * 2019-05-14 2020-11-19
CN112941039A (zh) * 2021-02-01 2021-06-11 南京大学 一种新型类囊泡溶瘤病毒及其在制备抗肿瘤药物上的应用

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998004726A1 (en) 1996-07-26 1998-02-05 Medical Research Council Hsv strain lacking functional icp27 and icp34.5 genes
WO1998051809A1 (en) 1997-05-12 1998-11-19 University Of Pittsburgh Of The Commonwealth System Of Higher Education Multigene vectors derived from hsv
WO1999060145A1 (en) 1998-05-20 1999-11-25 Biovex Limited Mutant herpes simplex viruses and uses thereof
WO2001053506A2 (en) 2000-01-21 2001-07-26 Biovex Limited Virus strains for the oncolytic treatment of cancer
US7820628B2 (en) 2005-02-22 2010-10-26 University Of Massachusetts Medical School Tumor lesion regression and conversion in situ into autologous tumor vaccines by compositions that result in anti-Gal antibody binding

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7338932B2 (en) * 2000-05-11 2008-03-04 Glycozym Aps Methods of modulating functions of polypeptide GalNAc-transferases and of screening test substances to find agents herefor, pharmaceutical compositions comprising such agents and the use of such agents for preparing medicaments
US20030099616A1 (en) * 2001-07-25 2003-05-29 Irving John M. Dual specificity tumor killing vectors driven by the telomerase promoter
CN1756568A (zh) * 2002-10-09 2006-04-05 衣阿华中央卫生系统 使用表达α(1,3)-半乳糖基转移酶的同种异型肿瘤细胞的抗肿瘤免疫
US20050201993A1 (en) * 2002-10-09 2005-09-15 Link Charles J.Jr. Antitumor vaccination using allogeneic tumor cells expressing alpha (1,3)-galactosyltransferase
EA013615B1 (ru) * 2004-11-12 2010-06-30 Байер Шеринг Фарма Акциенгезельшафт Рекомбинантный онколитический парамиксовирус и его применение
EP2085092A1 (en) * 2008-01-29 2009-08-05 Bayer Schering Pharma Aktiengesellschaft Attenuated oncolytic paramyxoviruses encoding avian cytokines
ES2385251B1 (es) * 2009-05-06 2013-05-06 Fundació Privada Institut D'investigació Biomèdica De Bellvitge Adenovirus oncolíticos para el tratamiento del cáncer.
EP2697368B1 (en) * 2011-04-15 2019-06-05 Genelux Corporation Clonal strains of attenuated vaccinia viruses and methods of use thereof
US9783789B2 (en) * 2012-12-07 2017-10-10 North Carolina State University Beta-hexosyl-transferases and uses thereof
CN105779397B (zh) * 2014-12-22 2019-07-19 彭霞 溶瘤异源重组新城疫病毒及其制备方法与应用

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998004726A1 (en) 1996-07-26 1998-02-05 Medical Research Council Hsv strain lacking functional icp27 and icp34.5 genes
WO1998051809A1 (en) 1997-05-12 1998-11-19 University Of Pittsburgh Of The Commonwealth System Of Higher Education Multigene vectors derived from hsv
WO1999060145A1 (en) 1998-05-20 1999-11-25 Biovex Limited Mutant herpes simplex viruses and uses thereof
WO2001053506A2 (en) 2000-01-21 2001-07-26 Biovex Limited Virus strains for the oncolytic treatment of cancer
US7820628B2 (en) 2005-02-22 2010-10-26 University Of Massachusetts Medical School Tumor lesion regression and conversion in situ into autologous tumor vaccines by compositions that result in anti-Gal antibody binding

Non-Patent Citations (45)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 2000, LIPPINCOTT, WILLIAMS & WILKINS
"Remington's Pharmaceutical Sciences", MEADE PUBLISHING CO.
ABDEL-MOTAL ET AL., CANCER IMMUNOL IMMUNOTHER., vol. 58, 2009, pages 1545 - 55
ANNIINA KOSKI ET AL: "Treatment of Cancer Patients With a Serotype 5/3 Chimeric Oncolytic Adenovirus Expressing GMCSF", MOLECULAR THERAPY, vol. 18, no. 10, 1 October 2010 (2010-10-01), pages 1874 - 1884, XP055015338, ISSN: 1525-0016, DOI: 10.1038/mt.2010.161 *
BRAMANTE ET AL., INT. J. CANCER, vol. 137, 2015, pages 1775 - 1783
DANTHINNE, BIOTECHNIQUES, vol. 30, no. 612-6, 2001, pages 618 - 9
DANTHINNE; WERTH, GENE THER., vol. 7, 2000, pages 80 - 7
DERIY ET AL., CANCER GENE THERAPY, vol. 12, 2005, pages 528 - 539
DERIY ET AL., CANCER GENE THERAPY, vol. 12, no. 6, 2005, pages 528 - 39
DERIY ET AL., GLYCOBIOLOGY, vol. 12, 2002, pages 135
DERIY ET AL., GLYCOBIOLOGY, vol. 12, 2002, pages 135 - 144
DERIY ET AL., GLYCOBIOLOGY, vol. 12, no. 2, 2002, pages 135 - 44
DUNN ET AL., NAT IMMUNOL, vol. 3, 2002, pages 991 - 8
FUEYO ET AL., ONCOGENE, vol. 19, 2000, pages 2 - 12
FUEYO J ET AL: "A mutant oncolytic adenovirus targeting the Rb pathway produces anti-glioma effect in vivo", ONCOGENE, NATURE PUBLISHING GROUP, GB, vol. 19, no. 1, 6 January 2000 (2000-01-06), pages 2 - 12, XP002680362, ISSN: 0950-9232 *
GALILI ET AL., J IMMUNOL., vol. 178, 2007, pages 4676 - 87
GALILI ET AL., TRANSPLANTATION, vol. 65, 1998, pages 1129 - 1132
GERLIER ET AL., TRENDS MICROBIOL., vol. 3, 1995, pages 338 - 345
HENION, T.R.; B.A. MACHER; F. ANARAKI; U. GALILI, GLYCOBIOLOGY, vol. 4, 1994, pages 193 - 201
KANERVA ET AL., CLIN. CANCER RES., vol. 19, 2013, pages 2734 - 2744
KANERVA ET AL., MOL. THER., vol. 8, 2003, pages 449 - 458
KANNO ET AL., ANTICANCER RES., vol. 32, 2012, pages 4891 - 5
KIM ET AL., GYNECOL. ONCOL., vol. 130, 2013, pages 518 - 24
KOSKI ET AL., MOL. THER., vol. 18, 2010, pages 1874 - 84
LIU ET AL., GENE THERAPY, vol. 10, no. 4, 2003, pages 292 - 303
LIU ET AL., NATURE CLINICAL PRACTICE ONCOLOGY, vol. 4, no. 2, 2007, pages 101 - 117
LIU ET AL., WORLD J GASTROENTEROL., vol. 19, no. 31, 21 August 2013 (2013-08-21), pages 5138 - 43
LUCY DERIY ET AL: "In vivo targeting of vaccinating tumor cells to antigen-presenting cells by a gene therapy method with adenovirus containing the [alpha]1,3galactosyltransferase gene", CANCER GENE THERAPY, vol. 12, no. 6, 8 April 2005 (2005-04-08), GB, pages 528 - 539, XP055279376, ISSN: 0929-1903, DOI: 10.1038/sj.cgt.7700812 *
LUGADE ET AL., J IMMUNOL., vol. 174, 2004, pages 7516 - 23
MALMBERG, CANCER IMMUNOL IMMUNOTHER., vol. 53, pages 879 - 92
MAXINE BAUZON ET AL: "Armed Therapeutic Viruses - A Disruptive Therapy on the Horizon of Cancer Immunotherapy", FRONTIERS IN IMMUNOLOGY, vol. 5, 1 January 2014 (2014-01-01), XP055214432, DOI: 10.3389/fimmu.2014.00074 *
NAKAMURA ET AL., EXPERT OPIN. BIO. THER., vol. 4, no. 10, 2004, pages 1685 - 1692
PARDOLL, CLIN IMMUNOL., vol. 95, 2000, pages 44 - 49
RAFIQ ET AL., J. CLIN. INVEST., vol. 110, 2002, pages 71 - 9
REED; MUENCH, AM. J. EPIDEMIOL., vol. 27, 1938, pages 493 - 497
REGNAULT ET AL., J. EXP. MED., vol. 189, 1999, pages 371 - 80
RUSSELL ET AL., VIROLOGY, vol. 199, 1994, pages 160 - 168
SAMBROOK ET AL.: "Molecular Cloning--A laboratory manual", 1989, COLD SPRING HARBOR PRESS
SCHAGEN ET AL., GENE THER., vol. 7, 2000, pages 1570 - 4
SCHREIBER ET AL., SCIENCE (NEW YORK, N. Y.), vol. 331, no. 6024, 2011, pages 1565 - 70
SHERR, SCIENCE, vol. 274, 1996, pages 1672 - 7
SIMONA BRAMANTE ET AL: "Treatment of melanoma with a serotype 5/3 chimeric oncolytic adenovirus coding for GM-CSF: Results in vitro , in rodents and in humans", INTERNATIONAL JOURNAL OF CANCER, vol. 137, no. 7, 26 March 2015 (2015-03-26), US, pages 1775 - 1783, XP055280194, ISSN: 0020-7136, DOI: 10.1002/ijc.29536 *
TATSUO ET AL., NATURE, vol. 406, no. 6798, 2000, pages 893 - 7
ULASOV ET AL., HUM. GENE THER., vol. 17, 2006, pages 556 - 64
URI GALILI: "Anti-Gal: an abundant human natural antibody of multiple pathogeneses and clinical benefits", IMMUNOLOGY., vol. 140, no. 1, 12 August 2013 (2013-08-12), GB, pages 1 - 11, XP055280049, ISSN: 0019-2805, DOI: 10.1111/imm.12110 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4087920A4 (en) * 2020-01-10 2024-02-28 Univ Hong Kong RECOMBINANT VIRUSES EXPRESSING ALPHA-1, 3-GALACTOSYLTRANSFERASE AND THEIR USES

Also Published As

Publication number Publication date
JP7066812B2 (ja) 2022-05-13
RU2720984C2 (ru) 2020-05-15
HK1251009B (zh) 2020-06-26
KR102544032B1 (ko) 2023-06-14
US20200155626A1 (en) 2020-05-21
CN107787364A (zh) 2018-03-09
BR112017021700A2 (pt) 2018-07-24
CA2981925A1 (en) 2016-10-13
IL254842A0 (en) 2017-12-31
GB201505860D0 (en) 2015-05-20
US20180104288A1 (en) 2018-04-19
EP3280798A1 (en) 2018-02-14
IL254842B (en) 2022-01-01
RU2017134519A (ru) 2019-04-04
RU2017134519A3 (ja) 2019-10-10
EP3280798B1 (en) 2019-06-19
KR20180015113A (ko) 2018-02-12
ES2743952T3 (es) 2020-02-21
MX2017012867A (es) 2018-05-11
JP2021045152A (ja) 2021-03-25
JP2018512150A (ja) 2018-05-17

Similar Documents

Publication Publication Date Title
EP3280798B1 (en) Therapeutic compositions of use for treating cancer
JP4588296B2 (ja) キメラワクチン
AU2011306845C1 (en) Oncolytic adenoviral vectors coding for monoclonal anti - CTLA - 4 antibodies
JP5284780B2 (ja) 癌の治療のための単純ヘルペスウイルス2型変異体の使用
US9744224B2 (en) Methods for treating cancer by administration of nucleic acids encoding FAP and cancer antigens
JP2019501671A (ja) 操作された腫瘍溶解性ウイルス
AU2014221143B2 (en) Vaccine composition
EP3256140B1 (en) Compositions for ebola virus vaccination
CN113456812A (zh) 用于联合免疫治疗的方法和组合物
JP2005521398A (ja) 癌治療用の強力な腫瘍溶解性単純ヘルペスウイルス
WO2005092374A2 (en) Recombinant herpes simplex virus and uses therefor
WO2005037321A1 (ja) 癌遺伝子治療薬
EP2494978A1 (en) Enhanced tumor therapy by tumor stem cell targeted oncolytic viruses
EP2563387B1 (en) Medical use of adenovirus vaccine vectors
Kirchhammer Tumor-targeted immunotherapy using an engineered adenoviral vector platform
FI124927B (fi) Adenovirusvektoreita ja niihin liittyviä menetelmiä ja käyttöjä

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16716896

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 20177027601

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 254842

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2017134519

Country of ref document: RU

ENP Entry into the national phase

Ref document number: 2981925

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2017552992

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 15564774

Country of ref document: US

Ref document number: MX/A/2017/012867

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112017021700

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112017021700

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20171009